Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
Add more filters

Publication year range
1.
J Biol Chem ; 295(2): 403-414, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31792057

ABSTRACT

The Plasmodium falciparum circumsporozoite protein (PfCSP) is a sporozoite surface protein whose role in sporozoite motility and cell invasion has made it the leading candidate for a pre-erythrocytic malaria vaccine. However, production of high yields of soluble recombinant PfCSP, including its extensive NANP and NVDP repeats, has proven problematic. Here, we report on the development and characterization of a secreted, soluble, and stable full-length PfCSP (containing 4 NVDP and 38 NANP repeats) produced in the Lactococcus lactis expression system. The recombinant full-length PfCSP, denoted PfCSP4/38, was produced initially with a histidine tag and purified by a simple two-step procedure. Importantly, the recombinant PfCSP4/38 retained a conformational epitope for antibodies as confirmed by both in vivo and in vitro characterizations. We characterized this complex protein by HPLC, light scattering, MS analysis, differential scanning fluorimetry, CD, SDS-PAGE, and immunoblotting with conformation-dependent and -independent mAbs, which confirmed it to be both pure and soluble. Moreover, we found that the recombinant protein is stable at both frozen and elevated-temperature storage conditions. When we used L. lactis-derived PfCSP4/38 to immunize mice, it elicited high levels of functional antibodies that had the capacity to modify sporozoite motility in vitro We concluded that the reported yield, purity, results of biophysical analyses, and stability of PfCSP4/38 warrant further consideration of using the L. lactis system for the production of circumsporozoite proteins for preclinical and clinical applications in malaria vaccine development.


Subject(s)
Lactococcus lactis/genetics , Malaria Vaccines/chemistry , Plasmodium falciparum/chemistry , Protozoan Proteins/chemistry , Animals , Cell Line , Female , Gene Expression , Humans , Malaria Vaccines/genetics , Malaria Vaccines/pharmacology , Malaria, Falciparum/prevention & control , Mice , Plasmodium falciparum/genetics , Protein Folding , Protein Stability , Protozoan Proteins/genetics , Protozoan Proteins/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Solubility
2.
Biochem Biophys Res Commun ; 534: 86-93, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33316544

ABSTRACT

This work describes a methodology for developing a minimal, subunit-based, multi-epitope, multi-stage, chemically-synthesised, anti-Plasmodium falciparum malaria vaccine. Some modified high activity binding peptides (mHABPs) derived from functionally relevant P. falciparum MSP, RH5 and AMA-1 conserved amino acid regions (cHABPs) for parasite binding to and invasion of red blood cells (RBC) were selected. They were highly immunogenic as assessed by indirect immunofluorescence (IFA) and Western blot (WB) assays and protective immune response-inducers against malarial challenge in the Aotus monkey experimental model. NetMHCIIpan 4.0 was used for predicting peptide-Aotus/human major histocompatibility class II (MHCII) binding affinity in silico due to the similarity between Aotus and human immune system molecules; ∼50% of Aotus MHCII allele molecules have a counterpart in the human immune system, being Aotus-specific, whilst others enabled recognition of their human counterparts. Some peptides' 1H-NMR-assessed structural conformation was determined to explain residue modifications in mHABPs inducing secondary structure changes. These directly influenced immunological behaviour, thereby highlighting the relationship with MHCII antigen presentation. The data obtained in such functional, immunological, structural and predictive approach suggested that some of these peptides could be excellent components of a fully-protective antimalarial vaccine.


Subject(s)
Erythrocytes/parasitology , Malaria Vaccines/pharmacology , Plasmodium falciparum/pathogenicity , Animals , Antigens, Protozoan/chemistry , Aotidae , Carrier Proteins/chemistry , Epitopes , Erythrocytes/drug effects , Histocompatibility Antigens Class II/metabolism , Host-Parasite Interactions/drug effects , Humans , Magnetic Resonance Spectroscopy , Malaria Vaccines/immunology , Malaria Vaccines/metabolism , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Peptides/immunology , Peptides/metabolism , Protozoan Proteins/chemistry , Vaccines, Subunit/immunology , Vaccines, Subunit/pharmacology
3.
PLoS Med ; 17(11): e1003377, 2020 11.
Article in English | MEDLINE | ID: mdl-33253211

ABSTRACT

BACKGROUND: The RTS,S/AS01 vaccine against Plasmodium falciparum malaria infection completed phase III trials in 2014 and demonstrated efficacy against clinical malaria of approximately 36% over 4 years for a 4-dose schedule in children aged 5-17 months. Pilot vaccine implementation has recently begun in 3 African countries. If the pilots demonstrate both a positive health impact and resolve remaining safety concerns, wider roll-out could be recommended from 2021 onwards. Vaccine demand may, however, outstrip initial supply. We sought to identify where vaccine introduction should be prioritised to maximise public health impact under a range of supply constraints using mathematical modelling. METHODS AND FINDINGS: Using a mathematical model of P. falciparum malaria transmission and RTS,S vaccine impact, we estimated the clinical cases and deaths averted in children aged 0-5 years in sub-Saharan Africa under 2 scenarios for vaccine coverage (100% and realistic) and 2 scenarios for other interventions (current coverage and World Health Organization [WHO] Global Technical Strategy targets). We used a prioritisation algorithm to identify potential allocative efficiency gains from prioritising vaccine allocation among countries or administrative units to maximise cases or deaths averted. If malaria burden at introduction is similar to current levels-assuming realistic vaccine coverage and country-level prioritisation in areas with parasite prevalence >10%-we estimate that 4.3 million malaria cases (95% credible interval [CrI] 2.8-6.8 million) and 22,000 deaths (95% CrI 11,000-35,000) in children younger than 5 years could be averted annually at a dose constraint of 30 million. This decreases to 3.0 million cases (95% CrI 2.0-4.7 million) and 14,000 deaths (95% CrI 7,000-23,000) at a dose constraint of 20 million, and increases to 6.6 million cases (95% CrI 4.2-10.8 million) and 38,000 deaths (95% CrI 18,000-61,000) at a dose constraint of 60 million. At 100% vaccine coverage, these impact estimates increase to 5.2 million cases (95% CrI 3.5-8.2 million) and 27,000 deaths (95% CrI 14,000-43,000), 3.9 million cases (95% CrI 2.7-6.0 million) and 19,000 deaths (95% CrI 10,000-30,000), and 10.0 million cases (95% CrI 6.7-15.7 million) and 51,000 deaths (95% CrI 25,000-82,000), respectively. Under realistic vaccine coverage, if the vaccine is prioritised sub-nationally, 5.3 million cases (95% CrI 3.5-8.2 million) and 24,000 deaths (95% CrI 12,000-38,000) could be averted at a dose constraint of 30 million. Furthermore, sub-national prioritisation would allow introduction in almost double the number of countries compared to national prioritisation (21 versus 11). If vaccine introduction is prioritised in the 3 pilot countries (Ghana, Kenya, and Malawi), health impact would be reduced, but this effect becomes less substantial (change of <5%) if 50 million or more doses are available. We did not account for within-country variation in vaccine coverage, and the optimisation was based on a single outcome measure, therefore this study should be used to understand overall trends rather than guide country-specific allocation. CONCLUSIONS: These results suggest that the impact of constraints in vaccine supply on the public health impact of the RTS,S malaria vaccine could be reduced by introducing the vaccine at the sub-national level and prioritising countries with the highest malaria incidence.


Subject(s)
Malaria Vaccines , Malaria, Falciparum/prevention & control , Malaria/prevention & control , Models, Theoretical , Child , Child, Preschool , Female , Ghana , Humans , Incidence , Infant , Infant, Newborn , Kenya , Malaria/epidemiology , Malaria Vaccines/administration & dosage , Malaria Vaccines/pharmacology , Malaria, Falciparum/epidemiology , Malawi , Male , Public Health/statistics & numerical data
4.
Cell Microbiol ; 21(7): e13030, 2019 07.
Article in English | MEDLINE | ID: mdl-30965383

ABSTRACT

An effective vaccine is a priority for malaria control and elimination. The leading candidate in the Plasmodium falciparum blood stage is PfRh5. PfRh5 assembles into trimeric complex with PfRipr and PfCyRPA in the parasite, and this complex is essential for erythrocyte invasion. In this study, we show that antibodies specific for PfRh5 and PfCyRPA prevent trimeric complex formation. We identify the EGF-7 domain on PfRipr as a neutralising epitope and demonstrate that antibodies against this region act downstream of complex formation to prevent merozoite invasion. Antibodies against the C-terminal region of PfRipr were more inhibitory than those against either PfRh5 or PfCyRPA alone, and a combination of antibodies against PfCyRPA and PfRipr acted synergistically to reduce invasion. This study supports prioritisation of PfRipr for development as part of a next-generation antimalarial vaccine.


Subject(s)
Antibodies, Neutralizing/pharmacology , Antigens, Protozoan/genetics , Carrier Proteins/genetics , Malaria, Falciparum/drug therapy , Protozoan Proteins/genetics , Antibodies, Neutralizing/immunology , Carrier Proteins/antagonists & inhibitors , Erythrocytes/drug effects , Erythrocytes/immunology , Humans , Malaria Vaccines/immunology , Malaria Vaccines/pharmacology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Merozoites/drug effects , Merozoites/immunology , Plasmodium falciparum/immunology , Plasmodium falciparum/pathogenicity , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/immunology
5.
Malar J ; 19(1): 421, 2020 Nov 23.
Article in English | MEDLINE | ID: mdl-33228666

ABSTRACT

To maintain momentum towards improved malaria control and elimination, a vaccine would be a key addition to the intervention toolkit. Two approaches are recommended: (1) promote the development and short to medium term deployment of first generation vaccine candidates and (2) support innovation and discovery to identify and develop highly effective, long-lasting and affordable next generation malaria vaccines.


Subject(s)
Biomedical Research , Drug Discovery/statistics & numerical data , Malaria Vaccines , Malaria Vaccines/analysis , Malaria Vaccines/chemistry , Malaria Vaccines/isolation & purification , Malaria Vaccines/pharmacology
6.
Malar J ; 18(1): 394, 2019 Dec 03.
Article in English | MEDLINE | ID: mdl-31796032

ABSTRACT

BACKGROUND: Despite the extensive endeavours, developing an effective malaria vaccine remains as a great challenge. Apical membrane antigen 1 (AMA-1) located on the merozoite surface of parasites belonging to the genus Plasmodium is involved in red blood cell invasion. METHODS: Influenza virus-like particle (VLP) vaccines containing codon-optimized or native (non-codon optimized) AMA-1 from Plasmodium berghei were generated. VLP-induced protective immunity was evaluated in a mouse model. RESULTS: Mice immunized with VLP vaccine containing the codon-optimized AMA-1 elicited higher levels of P. berghei-specific IgG and IgG2a antibody responses compared to VLPs containing non-codon optimized AMA-1 before and after challenge infection. Codon-optimized AMA-1 VLP vaccination induced higher levels of CD4+ T cells, CD8+ T cells, B cells, and germinal centre cell responses compared to non-codon optimized AMA-1 VLPs. Importantly, the codon-optimized AMA-1 VLP vaccination showed lower body weight loss, longer survival and a significant decrease in parasitaemia compared to non-codon optimized VLP vaccination. CONCLUSION: Overall, VLP vaccine expressing codon-optimized AMA-1 induced better protective efficacy than VLPs expressing the non-codon optimized AMA-1. Current findings highlight the importance of codon-optimization for vaccine use and its potential involvement in future malaria vaccine design strategies.


Subject(s)
Antigens, Protozoan/therapeutic use , Malaria Vaccines/pharmacology , Malaria/prevention & control , Membrane Proteins/therapeutic use , Plasmodium berghei/immunology , Protozoan Proteins/therapeutic use , Vaccines, Virus-Like Particle/pharmacology , Animals , Codon/immunology , Female , Mice , Mice, Inbred BALB C
7.
J Immunol ; 198(1): 300-307, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27903741

ABSTRACT

As a malaria transmission-blocking vaccine alone does not confer a direct benefit to the recipient, it is necessary to develop a vaccine that not only blocks malaria transmission but also protects vaccinated individuals. In this study we observed that a whole-killed blood-stage vaccine (WKV) not only conferred protection against the blood-stage challenge but also markedly inhibited the transmission of different strains of the malaria parasite. Although the parasitemia is much lower in WKV-immunized mice challenged with malaria parasites, the gametocytemia is comparable between control and immunized mice during the early stages of infection. The depletion of CD4+ T cells prior to the adoptive transfer of parasites into WKV-immunized mice has no effect on the development of the malaria parasite in the mosquito, but the adoptive transfer of the serum from the immunized mice into the parasite-inoculated mice remarkably suppresses the development of malaria parasites in mosquitoes. Furthermore, immunized mice challenged with the malaria parasite generate higher levels of parasite-specific Abs and the inflammatory cytokines MCP-1 and IFN-γ. However, the adoptive transfer of parasite-specific IgG or the depletion of MCP-1, but not IFN-γ, to some extent is closely associated with the suppression of malaria parasite development in mosquitoes. These data strongly suggest that WKV-induced immune responses confer protection against the mosquito stage, which is largely dependent on malaria parasite-specific Abs and MCP-1. This finding sheds new light on blocking malaria transmission through the immunization of individuals with the WKV.


Subject(s)
Culicidae/parasitology , Malaria Vaccines/immunology , Malaria/prevention & control , Malaria/transmission , Adoptive Transfer , Animals , Antibodies, Protozoan/immunology , Life Cycle Stages/drug effects , Malaria Vaccines/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Plasmodium yoelii/drug effects , Plasmodium yoelii/growth & development , Plasmodium yoelii/immunology , Vaccines, Inactivated/immunology , Vaccines, Inactivated/pharmacology
8.
BMC Med ; 16(1): 109, 2018 07 13.
Article in English | MEDLINE | ID: mdl-30001708

ABSTRACT

BACKGROUND: The RTS,S/AS01 vaccine for Plasmodium falciparum malaria demonstrated moderate efficacy in 5-17-month-old children in phase 3 trials, and from 2018, the vaccine will be evaluated through a large-scale pilot implementation program. Work is ongoing to optimise this vaccine, with higher efficacy for a different schedule demonstrated in a phase 2a challenge study. The objective of our study was to investigate the population-level impact of a modified RTS,S/AS01 schedule and dose amount in order to inform the target product profile for a second-generation malaria vaccine. METHODS: We used a mathematical modelling approach as the basis for our study. We simulated the changing anti-circumsporozoite antibody titre following vaccination and related the titre to vaccine efficacy. We then implemented this efficacy profile within an individual-based model of malaria transmission. We compared initial efficacy, duration and dose timing, and evaluated the potential public health impact of a modified vaccine in children aged 5-17 months, measuring clinical cases averted in children younger than 5 years. RESULTS: In the first decade of delivery, initial efficacy was associated with a higher reduction in childhood clinical cases compared to vaccine duration. This effect was more pronounced in high transmission settings and was due to the efficacy benefit occurring in younger ages where disease burden is highest. However, the low initial efficacy and long duration schedule averted more cases across all age cohorts if a longer time horizon was considered. We observed an age-shifting effect due to the changing immunological profile in higher transmission settings, in scenarios where initial efficacy was higher, and the fourth dose administered earlier. CONCLUSIONS: Our findings indicate that, for an imperfect childhood malaria vaccine with suboptimal efficacy, it may be advantageous to prioritise initial efficacy over duration. We predict that a modified vaccine could outperform the current RTS,S/AS01, although fourth dose timing will affect the age group that derives the greatest benefit. Further, the outcome measure and timeframe over which a vaccine is assessed are important when prioritising vaccine elements. This study provides insight into the most important characteristics of a malaria vaccine for at-risk groups and shows how distinct vaccine properties translate to public health outcomes. These findings may be used to prioritise target product profile elements for second-generation childhood malaria vaccines.


Subject(s)
Malaria Vaccines/therapeutic use , Malaria/prevention & control , Child , Child, Preschool , Female , Humans , Infant , Malaria Vaccines/pharmacology , Time Factors
9.
BMC Med ; 16(1): 61, 2018 04 30.
Article in English | MEDLINE | ID: mdl-29706136

ABSTRACT

BACKGROUND: Antibodies targeting Plasmodium falciparum sporozoites play a key role in human immunity to malaria. However, antibody mechanisms that neutralize sporozoites are poorly understood. This has been a major constraint in developing highly efficacious vaccines, as we lack strong correlates of protective immunity. METHODS: We quantified the ability of human antibodies from malaria-exposed populations to interact with human complement, examined the functional effects of complement activity against P. falciparum sporozoites in vitro, and identified targets of functional antibodies. In children and adults from malaria-endemic regions, we determined the acquisition of complement-fixing antibodies to sporozoites and their relationship with antibody isotypes and subclasses. We also investigated associations with protective immunity in a longitudinal cohort of children (n = 206) residing in a malaria-endemic region. RESULTS: We found that antibodies to the major sporozoite surface antigen, circumsporozoite protein (CSP), were predominately IgG1, IgG3, and IgM, and could interact with complement through recruitment of C1q and activation of the classical pathway. The central repeat region of CSP, included in leading vaccines, was a key target of complement-fixing antibodies. We show that antibodies activate human complement on P. falciparum sporozoites, which consequently inhibited hepatocyte cell traversal that is essential for establishing liver-stage infection, and led to sporozoite death in vitro. The natural acquisition of complement-fixing antibodies in malaria-exposed populations was age-dependent, and was acquired more slowly to sporozoite antigens than to merozoite antigens. In a longitudinal cohort of children, high levels of complement-fixing antibodies were significantly associated with protection against clinical malaria. CONCLUSIONS: These novel findings point to complement activation by antibodies as an important mechanism of anti-sporozoite human immunity, thereby enabling new strategies for developing highly efficacious malaria vaccines. We also present evidence that complement-fixing antibodies may be a valuable correlate of protective immunity in humans.


Subject(s)
Malaria Vaccines/therapeutic use , Malaria/prevention & control , Plasmodium falciparum/immunology , Sporozoites/immunology , Female , Humans , Malaria Vaccines/pharmacology , Male
10.
Bioconjug Chem ; 29(3): 761-770, 2018 03 21.
Article in English | MEDLINE | ID: mdl-28795800

ABSTRACT

As one of the pathogen-associated molecular patterns (PAMPs), flagellin is recently utilized as a potent adjuvant for many subunit vaccines. In this study, a truncated flagellin (tFL) with deletion of the hypervariable regions was adopted as a carrier-adjuvant by chemical conjugation with a chimeric malaria antigen M.RCAg-1 (M312) via a heterobifunctional polyethylene glycol (PEG) linker. After booster immunization in mice without any extra adjuvants, the M312-PEG-tFL conjugates elicited M312-specific antibody titers 100-1000 times higher than M312 and 10-100 times higher than the physical mixture of M312 and tFL. The elicited specific antibodies could recognize the native parasites, and the immunofluorescence assay (IFA) titer was 2100 for M312-P5k-tFL, which was about 7 times higher than M312. Furthermore, the IFA titers of the conjugates were comparable to the positive control of complete Freund's adjuvant (CFA). Compared to M312, the M312-PEG-tFL conjugates enhanced the proliferation index, lymphocyte activation, and memory T-cell generation. IgG subclasses of sera and cytokines analysis of splenocytes showed that conjugation with tFL could slightly trigger the Th1 polarization, while the antigen alone predominantly induced a Th2-biased immune response. Furthermore, a more-efficient innate immune response was provoked by the M312-PEG-tFL conjugates, as determined by the detection of antigen-specific TNF-α secretion by splenocytes. Our results indicated that tFL mainly retained the function as an agonist of TLR5. Conjugation of antigen to tFL could induce strong humoral and moderate cellular immune responses. Thus, conjugation of antigen to tFL as a potent carrier-adjuvant is an effective strategy for developing a promising protein-based vaccine.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Flagellin/therapeutic use , Immunity, Humoral , Malaria Vaccines/therapeutic use , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Vaccines, Conjugate/therapeutic use , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Animals , Antibody Formation , Flagellin/chemistry , Flagellin/pharmacology , Immunity, Cellular , Malaria Vaccines/chemistry , Malaria Vaccines/pharmacology , Malaria, Falciparum/immunology , Mice, Inbred BALB C , Vaccines, Conjugate/chemistry , Vaccines, Conjugate/pharmacology
11.
Parasitology ; 145(1): 56-70, 2018 01.
Article in English | MEDLINE | ID: mdl-27938428

ABSTRACT

The primate malaria Plasmodium knowlesi has a long-standing history as an experimental malaria model. Studies using this model parasite in combination with its various natural and experimental non-human primate hosts have led to important advances in vaccine development and in our understanding of malaria invasion, immunology and parasite-host interactions. The adaptation to long-term in vitro continuous blood stage culture in rhesus monkey, Macaca fascicularis and human red blood cells, as well as the development of various transfection methodologies has resulted in a highly versatile experimental malaria model, further increasing the potential of what was already a very powerful model. The growing evidence that P. knowlesi is an important human zoonosis in South-East Asia has added relevance to former and future studies of this parasite species.


Subject(s)
Disease Models, Animal , Haplorhini , Host-Parasite Interactions , Malaria/parasitology , Plasmodium knowlesi/physiology , Adaptation, Biological , Animals , Erythrocytes/parasitology , Humans , Macaca fascicularis , Macaca mulatta , Malaria/immunology , Malaria/prevention & control , Malaria/veterinary , Malaria Vaccines/analysis , Malaria Vaccines/pharmacology , Monkey Diseases/immunology , Monkey Diseases/parasitology , Monkey Diseases/prevention & control , Plasmodium knowlesi/immunology , Zoonoses/immunology , Zoonoses/parasitology , Zoonoses/prevention & control
12.
J Biol Chem ; 291(38): 19913-22, 2016 09 16.
Article in English | MEDLINE | ID: mdl-27432885

ABSTRACT

Development of a Plasmodium falciparum (Pf) transmission blocking vaccine (TBV) has the potential to significantly impact malaria control. Antibodies elicited against sexual stage proteins in the human bloodstream are taken up with the blood meal of the mosquitoes and inactivate parasite development in the mosquito. In a phase 1 trial, a leading TBV identified as Pfs25-EPA/Alhydrogel® appeared safe and immunogenic, however, the level of Pfs25-specific antibodies were likely too low for an effective vaccine. Pfs230, a 230-kDa sexual stage protein expressed in gametocytes is an alternative vaccine candidate. A unique 6-cysteine-rich domain structure within Pfs230 have thwarted its recombinant expression and characterization for clinical evaluation for nearly a quarter of a century. Here, we report on the identification, biochemical, biophysical, and immunological characterization of recombinant Pfs230 domains. Rabbit antibodies generated against recombinant Pfs230 domains blocked mosquito transmission of a laboratory strain and two field isolates using an ex vivo assay. A planned clinical trial of the Pfs230 vaccine is a significant step toward the potential development of a transmission blocking vaccine to eliminate malaria.


Subject(s)
Antigens, Protozoan/chemistry , Antigens, Protozoan/immunology , Malaria Vaccines/chemistry , Malaria Vaccines/immunology , Plasmodium falciparum/chemistry , Plasmodium falciparum/immunology , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Animals , Antibodies, Protozoan/immunology , Antigens, Protozoan/genetics , Antigens, Protozoan/pharmacology , Humans , Malaria Vaccines/genetics , Malaria Vaccines/pharmacology , Malaria, Falciparum/genetics , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Malaria, Falciparum/transmission , Plasmodium falciparum/genetics , Protein Domains , Protozoan Proteins/genetics , Protozoan Proteins/pharmacology , Rabbits
13.
PLoS Med ; 14(11): e1002455, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29190291

ABSTRACT

Since the turn of the century, a remarkable expansion has been achieved in the range and effectiveness of products and strategies available to prevent, treat, and control malaria, including advances in diagnostics, drugs, vaccines, and vector control. These advances have once again put malaria elimination on the agenda. However, it is clear that even with the means available today, malaria control and elimination pose a formidable challenge in many settings. Thus, currently available resources must be used more effectively, and new products and approaches likely to achieve these goals must be developed. This paper considers tools (both those available and others that may be required) to achieve and maintain malaria elimination. New diagnostics are needed to direct treatment and detect transmission potential; new drugs and vaccines to overcome existing resistance and protect against clinical and severe disease, as well as block transmission and prevent relapses; and new vector control measures to overcome insecticide resistance and more powerfully interrupt transmission. It is also essential that strategies for combining new and existing approaches are developed for different settings to maximise their longevity and effectiveness in areas with continuing transmission and receptivity. For areas where local elimination has been recently achieved, understanding which measures are needed to maintain elimination is necessary to prevent rebound and the reestablishment of transmission. This becomes increasingly important as more countries move towards elimination.


Subject(s)
Antimalarials/therapeutic use , Biomedical Research/methods , Insect Vectors/drug effects , Malaria Vaccines/therapeutic use , Malaria/prevention & control , Mosquito Control/methods , Animals , Antimalarials/pharmacology , Biomedical Research/trends , Disease Eradication/methods , Humans , Malaria/epidemiology , Malaria Vaccines/pharmacology , Mosquito Control/trends
14.
Proc Natl Acad Sci U S A ; 111(34): 12307-12, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-25136089

ABSTRACT

With the 2010s declared the Decade of Vaccines, and Millennium Development Goals 4 and 5 focused on reducing diseases that are potentially vaccine preventable, now is an exciting time for vaccines against poverty, that is, vaccines against diseases that disproportionately affect low- and middle-income countries (LMICs). The Global Burden of Disease Study 2010 has helped better understand which vaccines are most needed. In 2012, US$1.3 billion was spent on research and development for new vaccines for neglected infectious diseases. However, the majority of this went to three diseases: HIV/AIDS, malaria, and tuberculosis, and not neglected diseases. Much of it went to basic research rather than development, with an ongoing decline in funding for product development partnerships. Further investment in vaccines against diarrheal diseases, hepatitis C, and group A Streptococcus could lead to a major health impact in LMICs, along with vaccines to prevent sepsis, particularly among mothers and neonates. The Advanced Market Commitment strategy of the Global Alliance for Vaccines and Immunisation (GAVI) Alliance is helping to implement vaccines against rotavirus and pneumococcus in LMICs, and the roll out of the MenAfriVac meningococcal A vaccine in the African Meningitis Belt represents a paradigm shift in vaccines against poverty: the development of a vaccine primarily targeted at LMICs. Global health vaccine institutes and increasing capacity of vaccine manufacturers in emerging economies are helping drive forward new vaccines for LMICs. Above all, partnership is needed between those developing and manufacturing LMIC vaccines and the scientists, health care professionals, and policy makers in LMICs where such vaccines will be implemented.


Subject(s)
Vaccines/pharmacology , AIDS Vaccines/pharmacology , Communicable Disease Control/methods , Developing Countries , Diarrhea/prevention & control , Global Health , Humans , Malaria Vaccines/pharmacology , Neglected Diseases/prevention & control , Poverty Areas , Respiratory Tract Infections/prevention & control , Tuberculosis Vaccines/pharmacology , Vaccines/economics
15.
Proc Natl Acad Sci U S A ; 111(28): 10311-6, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24958881

ABSTRACT

An essential step in the invasion of red blood cells (RBCs) by Plasmodium falciparum (Pf) merozoites is the binding of rhoptry neck protein 2 (RON2) to the hydrophobic groove of apical membrane antigen 1 (AMA1), triggering junction formation between the apical end of the merozoite and the RBC surface to initiate invasion. Vaccination with AMA1 provided protection against homologous parasites in one of two phase 2 clinical trials; however, despite its ability to induce high-titer invasion-blocking antibodies in a controlled human challenge trial, the vaccine conferred little protection even against the homologous parasite. Here we provide evidence that immunization with an AMA1-RON2 peptide complex, but not with AMA1 alone, provided complete protection against a lethal Plasmodium yoelii challenge in mice. Significantly, IgG from mice immunized with the complex transferred protection. Furthermore, IgG from PfAMA1-RON2-immunized animals showed enhanced invasion inhibition compared with IgG elicited by AMA1 alone. Interestingly, this qualitative increase in inhibitory activity appears to be related, at least in part, to a switch in the proportion of IgG specific for certain loop regions in AMA1 surrounding the binding site of RON2. Antibodies induced by the complex were not sufficient to block the FVO strain heterologous parasite, however, reinforcing the need to include multiallele AMA1 to cover polymorphisms. Our results suggest that AMA1 subunit vaccines may be highly effective when presented to the immune system as an invasion complex with RON2.


Subject(s)
Antigens, Protozoan/pharmacology , Erythrocytes/immunology , Immunization , Malaria Vaccines/pharmacology , Malaria, Falciparum/immunology , Membrane Proteins/pharmacology , Multiprotein Complexes/pharmacology , Plasmodium falciparum/immunology , Protozoan Proteins/pharmacology , Animals , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Erythrocytes/parasitology , Humans , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Malaria, Falciparum/genetics , Malaria, Falciparum/prevention & control , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Multiprotein Complexes/genetics , Multiprotein Complexes/immunology , Plasmodium falciparum/genetics , Plasmodium yoelii/genetics , Plasmodium yoelii/immunology , Protozoan Proteins/genetics , Protozoan Proteins/immunology
16.
Bull World Health Organ ; 94(9): 640-641, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27708468

ABSTRACT

For the first time a malaria vaccine is to be tested for possible inclusion in national immunization programmes. Malcolm Molyneux tells Fiona Fleck why governments may need to work hard to convince people of its benefits.


Subject(s)
Malaria Vaccines/pharmacology , Malaria/prevention & control , Africa/epidemiology , Animals , Anopheles/parasitology , Clinical Trials as Topic , Global Health , Humans , Malaria/epidemiology , Malawi , Plasmodium falciparum/drug effects , World Health Organization
17.
J Immunol ; 193(5): 2073-86, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25080483

ABSTRACT

Polyvalent vaccines use a mixture of Ags representing distinct pathogen strains to induce an immune response that is cross-reactive and protective. However, such approaches often have mixed results, and it is unclear how polyvalency alters the fine specificity of the Ab response and what those consequences might be for protection. In this article, we present a coarse-grain theoretical model of B cell affinity maturation during monovalent and polyvalent vaccinations that predicts the fine specificity and cross-reactivity of the Ab response. We stochastically simulate affinity maturation using a population dynamics approach in which the host B cell repertoire is represented explicitly, and individual B cell subpopulations undergo rounds of stimulation, mutation, and differentiation. Ags contain multiple epitopes and are present in subpopulations of distinct pathogen strains, each with varying degrees of cross-reactivity at the epitope level. This epitope- and strain-specific model of affinity maturation enables us to study the composition of the polyclonal response in granular detail and identify the mechanisms driving serum specificity and cross-reactivity. We applied this approach to predict the Ab response to a polyvalent vaccine based on the highly polymorphic malaria Ag apical membrane antigen-1. Our simulations show how polyvalent apical membrane Ag-1 vaccination alters the selection pressure during affinity maturation to favor cross-reactive B cells to both conserved and strain-specific epitopes and demonstrate how a polyvalent vaccine with a small number of strains and only moderate allelic coverage may be broadly neutralizing. Our findings suggest that altered fine specificity and enhanced cross-reactivity may be a universal feature of polyvalent vaccines.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , B-Lymphocytes/microbiology , Malaria Vaccines/immunology , Membrane Proteins/immunology , Models, Immunological , Protozoan Proteins/immunology , Antigens, Protozoan/pharmacology , Cross Reactions , Humans , Malaria Vaccines/pharmacology , Membrane Proteins/pharmacology , Protozoan Proteins/pharmacology
18.
Malar J ; 14: 150, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25890243

ABSTRACT

BACKGROUND: A vaccine that interrupts malaria transmission (VIMT) would be a valuable tool for malaria control and elimination. One VIMT approach is to identify sexual erythrocytic and mosquito stage antigens of the malaria parasite that induce immune responses targeted at disrupting parasite development in the mosquito. The standard Plasmodium falciparum membrane-feeding assay (SMFA) is used to assess transmission-blocking activity (TBA) of antibodies against candidate immunogens and of drugs targeting the mosquito stages. To develop its P. falciparum sporozoite (SPZ) products, Sanaria has industrialized the production of P. falciparum-infected Anopheles stephensi mosquitoes, incorporating quantitative analyses of oocyst and P. falciparum SPZ infections as part of the manufacturing process. METHODS: These capabilities were exploited to develop a robust, reliable, consistent SMFA that was used to assess 188 serum samples from animals immunized with the candidate vaccine immunogen, Pfs25, targeting P. falciparum mosquito stages. Seventy-four independent SMFAs were performed. Infection intensity (number of oocysts/mosquito) and infection prevalence (percentage of mosquitoes infected with oocysts) were compared between mosquitoes fed cultured gametocytes plus normal human O(+) serum (negative control), anti-Pfs25 polyclonal antisera (MRA39 or MRA38, at a final dilution in the blood meal of 1:54 as positive control), and test sera from animals immunized with Pfs25 (at a final dilution in the blood meal of 1:9). RESULTS: SMFA negative controls consistently yielded high infection intensity (mean = 46.1 oocysts/midgut, range of positives 3.7-135.6) and infection prevalence (mean = 94.2%, range 71.4-100.0) and in positive controls, infection intensity was reduced by 81.6% (anti-Pfs25 MRA39) and 97.0% (anti-Pfs25 MRA38), and infection prevalence was reduced by 12.9 and 63.5%, respectively. A range of TBAs was detected among the 188 test samples assayed in duplicate. Consistent administration of infectious gametocytes to mosquitoes within and between assays was achieved, and the TBA of anti-Pfs25 control antibodies was highly reproducible. CONCLUSIONS: These results demonstrate a robust capacity to perform the SMFA in a medium-to-high throughput format, suitable for assessing large numbers of experimental samples of candidate antibodies or drugs.


Subject(s)
Anopheles/physiology , Antimalarials/pharmacology , Biological Assay/methods , Malaria Vaccines/pharmacology , Plasmodium falciparum/drug effects , Plasmodium falciparum/immunology , Animals , Feeding Behavior , Female , Membranes/physiology
19.
Gene Ther ; 21(2): 225-32, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24385146

ABSTRACT

In DNA vaccination, CD4(+) T-cell help can be enhanced by fusion of a gene encoding an immunization protein with a foreign gene or its part providing T(h) epitopes. To study the effect of helper epitope localization in a protein molecule, the influence of the vicinity of the helper epitope, and the impact of chimeric protein cellular localization, we fused the helper epitope p30 from tetanus toxin (TT, aa 947-967) with the N- or C-terminus of the mutated E7 oncoprotein (E7GGG) of human papillomavirus type 16, enlarged the p30 epitope with the flanking residues containing potential protease-sensitive sites and altered the cellular localization of the fusion constructs by signal sequences. The p30 epitope enhanced the E7-specific response, but only in constructs without added signal sequences. After localization of the fusion proteins into the endoplasmic reticulum and endo/lysosomal compartment, the TT-specific T(h)2 response was increased. The synthetic Pan DR epitope (PADRE) induced a stronger E7-specific response than the p30 epitope and its stimulatory effect was not limited to nuclear/cytoplasmic localization of the E7 antigen. These results suggest that in the optimization of immune responses by adding helper epitopes to DNA vaccines delivered by the gene gun, the cellular localization of the antigen needs to be taken into account.


Subject(s)
Biolistics/methods , Endoplasmic Reticulum/immunology , Malaria Vaccines/pharmacology , Papillomavirus E7 Proteins/genetics , Peptide Fragments/genetics , Tetanus Toxin/genetics , Vaccines, DNA/pharmacology , Animals , Cell Line, Tumor , Cytokines/metabolism , Endoplasmic Reticulum/metabolism , Female , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Papillomavirus E7 Proteins/metabolism , Papillomavirus E7 Proteins/pharmacology , Peptide Fragments/pharmacology , Plasmids/administration & dosage , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Tetanus Toxin/pharmacology , Vaccines, DNA/administration & dosage
20.
Biochem Biophys Res Commun ; 451(1): 15-23, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25063026

ABSTRACT

Developing novel generations of subunit-based antimalarial vaccines in the form of chemically-defined macromolecule systems for multiple antigen presentation represents a classical problem in the field of vaccine development. Many efforts involving synthesis strategies leading to macromolecule constructs have been based on dendrimer-like systems, the condensation of large building blocks and conventional asymmetric double dimer constructs, all based on lysine cores. This work describes novel symmetric double dimer and condensed linear constructs for presenting selected peptide multi-copies from the apical sushi protein expressed in Plasmodium falciparum. These molecules have been proved to be safe and innocuous, highly antigenic and have shown strong protective efficacy in rodents challenged with two Plasmodium species. Insights into systematic design, synthesis and characterisation have led to such novel antigen systems being used as potential platforms for developing new anti-malarial vaccine candidates.


Subject(s)
Antigens, Protozoan/chemistry , Malaria Vaccines/chemistry , Malaria Vaccines/pharmacology , Plasmodium falciparum/chemistry , Amino Acid Sequence , Aminocaproates/chemistry , Animals , Antigens, Protozoan/immunology , Antigens, Protozoan/metabolism , Epitopes , Humans , Malaria/prevention & control , Malaria, Falciparum/prevention & control , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Sequence Data , Peptides/immunology , Plasmodium berghei/pathogenicity , Plasmodium yoelii/pathogenicity , Protein Conformation , Protein Multimerization , Rabbits , Vaccines, Subunit/chemistry , Vaccines, Subunit/immunology
SELECTION OF CITATIONS
SEARCH DETAIL